Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Neurochem ; 168(2): 100-114, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38102893

RESUMO

The aquaporin-4 (AQP4) water channel is abundantly expressed in the glial cells of the central nervous system and facilitates brain swelling following diverse insults, such as traumatic injury or stroke. Lack of specific and therapeutic AQP4 inhibitors highlights the need to explore alternative routes to control the water permeability of glial cell membranes. The cell surface abundance of AQP4 in mammalian cells fluctuates rapidly in response to changes in oxygen levels and tonicity, suggesting a role for vesicular trafficking in its translocation to and from the cell surface. However, the molecular mechanisms of AQP4 trafficking are not fully elucidated. In this work, early and recycling endosomes were investigated as likely candidates of rapid AQP4 translocation together with changes in cytoskeletal dynamics. In transiently transfected HEK293 cells a significant amount of AQP-eGFP colocalised with mCherry-Rab5-positive early endosomes and mCherry-Rab11-positive recycling endosomes. When exposed to hypotonic conditions, AQP4-eGFP rapidly translocated from intracellular vesicles to the cell surface. Co-expression of dominant negative forms of the mCherry-Rab5 and -Rab11 with AQP4-eGFP prevented hypotonicity-induced AQP4-eGFP trafficking and led to concentration at the cell surface or intracellular vesicles respectively. Use of endocytosis inhibiting drugs indicated that AQP4 internalisation was dynamin-dependent. Cytoskeleton dynamics-modifying drugs also affected AQP4 translocation to and from the cell surface. AQP4 trafficking mechanisms were validated in primary human astrocytes, which express high levels of endogenous AQP4. The results highlight the role of early and recycling endosomes and cytoskeletal dynamics in AQP4 translocation in response to hypotonic and hypoxic stress and suggest continuous cycling of AQP4 between intracellular vesicles and the cell surface under physiological conditions.


Assuntos
Endocitose , Endossomos , Animais , Humanos , Células HEK293 , Transporte Proteico , Endossomos/metabolismo , Astrócitos/metabolismo , Aquaporina 4/genética , Aquaporina 4/metabolismo , Mamíferos/metabolismo
2.
Neurobiol Dis ; 185: 106236, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37495179

RESUMO

Huntington's disease (HD) is a fatal neurodegenerative disease with limited treatment options. Human and animal studies have suggested that metabolic and mitochondrial dysfunctions contribute to HD pathogenesis. Here, we use high-resolution respirometry to uncover defective mitochondrial oxidative phosphorylation and electron transfer capacity when a mutant huntingtin fragment is targeted to neurons or muscles in Drosophila and find that enhancing mitochondrial function can ameliorate these defects. In particular, we find that co-expression of parkin, an E3 ubiquitin ligase critical for mitochondrial dynamics and homeostasis, produces significant enhancement of mitochondrial respiration when expressed either in neurons or muscles, resulting in significant rescue of neurodegeneration, viability and longevity in HD model flies. Targeting mutant HTT to muscles results in larger mitochondria and higher mitochondrial mass, while co-expression of parkin increases mitochondrial fission and decreases mass. Furthermore, directly addressing HD-mediated defects in the fly's mitochondrial electron transport system, by rerouting electrons to either bypass mitochondrial complex I or complexes III-IV, significantly increases mitochondrial respiration and results in a striking rescue of all phenotypes arising from neuronal mutant huntingtin expression. These observations suggest that bypassing impaired mitochondrial respiratory complexes in HD may have therapeutic potential for the treatment of this devastating disorder.


Assuntos
Doença de Huntington , Doenças Neurodegenerativas , Animais , Humanos , Drosophila/metabolismo , Doenças Neurodegenerativas/metabolismo , Neurônios/metabolismo , Mitocôndrias/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Doença de Huntington/metabolismo , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo
3.
Respir Res ; 23(1): 183, 2022 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-35831901

RESUMO

BACKGROUND: Airway remodeling is a significant contributor to impaired lung function in chronic allergic airway disease. Currently, no therapy exists that is capable of targeting these structural changes and the consequent loss of function. In the context of chronic allergic inflammation, pericytes have been shown to uncouple from the pulmonary microvasculature, migrate to areas of inflammation, and significantly contribute to airway wall remodeling and lung dysfunction. This study aimed to elucidate the mechanism by which pulmonary pericytes accumulate in the airway wall in a model of chronic allergic airway inflammation. METHODS: Mice were subjected to a protocol of chronic airway inflammation driven by the common environmental aeroallergen house dust mite. Phenotypic changes to lung pericytes were assessed by flow cytometry and immunostaining, and the functional capacity of these cells was evaluated using in vitro migration assays. The molecular mechanisms driving these processes were targeted pharmacologically in vivo and in vitro. RESULTS: Pericytes demonstrated increased CXCR4 expression in response to chronic allergic inflammation and migrated more readily to its cognate chemokine, CXCL12. This increase in migratory capacity was accompanied by pericyte accumulation in the airway wall, increased smooth muscle thickness, and symptoms of respiratory distress. Pericyte uncoupling from pulmonary vessels and subsequent migration to the airway wall were abrogated following topical treatment with the CXCL12 neutraligand LIT-927. CONCLUSION: These results provide new insight into the role of the CXCL12/CXCR4 signaling axis in promoting pulmonary pericyte accumulation and airway remodeling and validate a novel target to address tissue remodeling associated with chronic inflammation.


Assuntos
Asma , Quimiocina CXCL12/metabolismo , Hipersensibilidade , Transtornos Respiratórios , Remodelação das Vias Aéreas , Animais , Modelos Animais de Doenças , Hipersensibilidade/metabolismo , Inflamação/metabolismo , Pulmão , Camundongos , Pericitos/metabolismo , Transtornos Respiratórios/metabolismo
4.
Hum Mol Genet ; 23(12): 3129-37, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24452335

RESUMO

Huntington's disease (HD) is a devastating neurodegenerative disorder which is inherited in an autosomal dominant manner. HD is caused by a trinucleotide CAG repeat expansion that encodes a polyglutamine stretch in the huntingtin (HTT) protein. Mutant HTT expression leads to a myriad of cellular dysfunctions culminating in neuronal loss and consequent motor, cognitive and psychiatric disturbances in HD patients. The length of the CAG repeat is inversely correlated with age of onset (AO) in HD patients, while environmental and genetic factors can further modulate this parameter. Here, we explored whether the recently described copy-number variation (CNV) of the gene SLC2A3-which encodes the neuronal glucose transporter GLUT3-could modulate AO in HD. Strikingly, we found that increased dosage of SLC2A3 delayed AO in an HD cohort of 987 individuals, and that this correlated with increased levels of GLUT3 in HD patient cells. To our knowledge this is the first time that CNV of a candidate gene has been found to modulate HD pathogenesis. Furthermore, we found that increasing dosage of Glut1-the Drosophila melanogaster homologue of this glucose transporter-ameliorated HD-relevant phenotypes in fruit flies, including neurodegeneration and life expectancy. As alterations in glucose metabolism have been implicated in HD pathogenesis, this study may have important therapeutic relevance for HD.


Assuntos
Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Transportador de Glucose Tipo 3/genética , Transportador de Glucose Tipo 3/metabolismo , Doença de Huntington/epidemiologia , Doença de Huntington/genética , Idade de Início , Animais , Linhagem Celular , Estudos de Coortes , Variações do Número de Cópias de DNA , Modelos Animais de Doenças , Feminino , Dosagem de Genes , Humanos , Doença de Huntington/patologia , Masculino , Filogenia , Regulação para Cima
5.
Biomolecules ; 13(8)2023 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-37627296

RESUMO

Whilst S100P has been shown to be a marker for carcinogenesis, we have shown, in non-physio-pathological states, that its expression promotes trophoblast motility and invasion but the mechanisms explaining these cellular processes are unknown. Here we identify the presence of S100P in the plasma membrane/cell surface of all trophoblast cells tested, whether lines, primary extravillous (EVT) cells, or section tissue samples using either biochemical purification of plasma membrane material, cell surface protein isolation through biotinylation, or microscopy analysis. Using extracellular loss of function studies, through addition of a specific S100P antibody, our work shows that inhibiting the cell surface/membrane-bound or extracellular S100P pools significantly reduces, but importantly only in part, both cell motility and cellular invasion in different trophoblastic cell lines, as well as primary EVTs. Interestingly, this loss in cellular motility/invasion did not result in changes to the overall actin organisation and focal adhesion complexes. These findings shed new light on at least two newly characterized pathways by which S100P promotes trophoblast cellular motility and invasion. One where cellular S100P levels involve the remodelling of focal adhesions whilst another, an extracellular pathway, appears to be focal adhesion independent. Both pathways could lead to the identification of novel targets that may explain why significant numbers of confirmed human pregnancies suffer complications through poor placental implantation.


Assuntos
Placenta , Trofoblastos , Feminino , Gravidez , Humanos , Membranas , Membrana Celular , Proteínas de Membrana , Anticorpos , Proteínas de Ligação ao Cálcio , Proteínas de Neoplasias
6.
Cells ; 12(6)2023 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-36980245

RESUMO

The c-Jun N-terminal kinases (JNKs) are a family of proteins that, once activated by stress stimuli, can alter neuronal functions and survival. The JNK cascade plays a crucial role in the post-synaptic neuronal compartment by altering its structural organization and leading, at worst, to an overall impairment of neuronal communication. Increasing evidence suggests that synaptic impairment is the first neurodegenerative event in Alzheimer's disease (AD). To better elucidate this mechanism, we longitudinally studied 5xFAD mice at three selected time points representative of human AD symptom progression. We tested the mice cognitive performance by using the radial arm water maze (RAWM) in parallel with biochemical evaluations of post-synaptic enriched protein fraction and total cortical parenchyma. We found that 5xFAD mice presented a strong JNK activation at 3.5 months of age in the post-synaptic enriched protein fraction. This JNK activation correlates with a structural alteration of the post-synaptic density area and with memory impairment at this early stage of the disease that progressively declines to cause cell death. These findings pave the way for future studies on JNK as a key player in early neurodegeneration and as an important therapeutic target for the development of new compounds able to tackle synaptic impairment in the early phase of AD pathology.


Assuntos
Doença de Alzheimer , Disfunção Cognitiva , Proteínas Quinases JNK Ativadas por Mitógeno , Animais , Camundongos , Doença de Alzheimer/metabolismo , Disfunção Cognitiva/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Sistema de Sinalização das MAP Quinases , Fosforilação , Modelos Animais de Doenças
7.
Neurobiol Dis ; 46(3): 710-21, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22426389

RESUMO

Limiting the development of secondary damage represents one of the major goals of neuroprotective therapies after spinal cord injury. Here, we demonstrate that specific JNK inhibition via a single intraperitoneal injection of the cell permeable peptide D-JNKI1 6h after lesion improves locomotor recovery assessed by both the footprint and the BMS tests up to 4 months post-injury in mice. JNK inhibition prevents c-jun phosphorylation and caspase-3 cleavage, has neuroprotective effects and results in an increased sparing of white matter at the lesion site. Lastly, D-JNKI1 treated animals show a lower increase of erythrocyte extravasation and blood brain barrier permeability, thus indicating protection of the vascular system. In total, these results clearly point out JNK inhibition as a promising neuroprotective strategy for preventing the evolution of secondary damage after spinal cord injury.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Locomoção/efeitos dos fármacos , Fármacos Neuroprotetores , Peptídeos/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Recuperação de Função Fisiológica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Traumatismos da Medula Espinal/tratamento farmacológico , Animais , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/fisiologia , Western Blotting , Caspase 3/metabolismo , Membro Posterior/fisiologia , Processamento de Imagem Assistida por Computador , Imageamento Tridimensional , Imuno-Histoquímica , Injeções Intraperitoneais , Masculino , Camundongos , Fibras Nervosas/fisiologia , Inibidores de Proteínas Quinases/administração & dosagem , Proteínas Proto-Oncogênicas c-jun/metabolismo , Serotonina/fisiologia , Medula Espinal/patologia , Traumatismos da Medula Espinal/enzimologia , Traumatismos da Medula Espinal/fisiopatologia
8.
Biomedicines ; 10(9)2022 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-36140394

RESUMO

The flavoprotein kynurenine 3-monooxygenase (KMO) is localised to the outer mitochondrial membrane and catalyses the synthesis of 3-hydroxykynurenine from L-kynurenine, a key step in the kynurenine pathway (KP) of tryptophan degradation. Perturbation of KP metabolism due to inflammation has long been associated with the pathogenesis of several neurodegenerative disorders, including Huntington's disease (HD)-which is caused by the expansion of a polyglutamine stretch in the huntingtin (HTT) protein. While HTT is primarily localised to the cytoplasm, it also associates with mitochondria, where it may physically interact with KMO. In order to test this hypothesis, we employed bimolecular fluorescence complementation (BiFC) and found that KMO physically interacts with soluble HTT exon 1 protein fragment in living cells. Notably, expansion of the disease-causing polyglutamine tract in HTT leads to the formation of proteinaceous intracellular inclusions that disrupt this interaction with KMO, markedly decreasing BiFC efficiency. Using confocal microscopy and ultrastructural analysis, we determined KMO and HTT localisation within the cell and found that the KMO-HTT interaction is localized to the outer mitochondrial membrane. These data suggest that KMO may interact with a pool of HTT at the mitochondrial membrane, highlighting a possible physiological role for mitochondrial HTT. The KMO-HTT interaction is abrogated upon polyglutamine expansion, which may indicate a heretofore unrecognized relevance in the pathogenesis of this disorder.

9.
Cerebellum ; 10(2): 281-90, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21191679

RESUMO

Several studies have shown that Purkinje cells die by apoptosis in organotypic slice cultures from postnatal 3-day-old (P3) mice. This cell death is age-dependent and has been proposed as indirect evidence for the programmed Purkinje cell death occurring in in vivo cerebellum. Here, we studied whether c-jun N-terminal kinase (JNK) and p38 kinase pathways contribute to the Purkinje cell death observed in cerebellar slice cultures obtained from P3 mice. Slice culture treatment with D-JNKI1 or SB203580, respectively inhibitors of JNK and p38 MAP kinases, results in a better survival of Purkinje cells. Interestingly, the combined treatment with the two inhibitors potentiated single treatment effects. These results suggest that p38 and JNK pathways might be differently implicated in this Purkinje cell death. Time course experiments found p38 activation immediately post-slicing, whereas JNK activation was detected only 2 h after the culture. We hypothesize that p38 activation might be due to the "sliced condition," and JNK activation might be more specific to P3 age-dependent cell death. The study of JNK and p38 activation in cerebellar lysates from P0 slice culture confirmed JNK activation being specific for the P3 explants, whereas p38 is activated both from P0 and P3 cerebellar slice culture lysates. These results suggest that p38 is activated by the slicing, whereas JNK activation is related to developmental Purkinje cell death.


Assuntos
Apoptose/fisiologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Células de Purkinje/citologia , Células de Purkinje/enzimologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Envelhecimento/fisiologia , Animais , Artefatos , Western Blotting , Ativação Enzimática , Camundongos , Técnicas de Cultura de Órgãos , Manejo de Espécimes/efeitos adversos
10.
Nat Med ; 9(9): 1180-6, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12937412

RESUMO

Neuronal death in cerebral ischemia is largely due to excitotoxic mechanisms, which are known to activate the c-Jun N-terminal kinase (JNK) pathway. We have evaluated the neuroprotective power of a cell-penetrating, protease-resistant peptide that blocks the access of JNK to many of its targets. We obtained strong protection in two models of middle cerebral artery occlusion (MCAO): transient occlusion in adult mice and permanent occlusion in 14-d-old rat pups. In the first model, intraventricular administration as late as 6 h after occlusion reduced the lesion volume by more than 90% for at least 14 d and prevented behavioral consequences. In the second model, systemic delivery reduced the lesion by 78% and 49% at 6 and 12 h after ischemia, respectively. Protection correlated with prevention of an increase in c-Jun activation and c-Fos transcription. In view of its potency and long therapeutic window, this protease-resistant peptide is a promising neuroprotective agent for stroke.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Isquemia Encefálica/prevenção & controle , Inibidores Enzimáticos/farmacologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Peptídeos/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Isquemia Encefálica/patologia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Células Cultivadas , Agonistas de Aminoácidos Excitatórios/efeitos adversos , Produtos do Gene tat/genética , Produtos do Gene tat/metabolismo , Genes fos/efeitos dos fármacos , Técnicas In Vitro , Infarto da Artéria Cerebral Média/tratamento farmacológico , Infarto da Artéria Cerebral Média/patologia , Proteínas Quinases JNK Ativadas por Mitógeno , Masculino , Camundongos , Camundongos Endogâmicos ICR , N-Metilaspartato/efeitos adversos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Ratos , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacologia
11.
J Neurosci ; 29(32): 10144-52, 2009 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-19675248

RESUMO

Presenilin 1 (PS1) mutations are responsible for a majority of early onset familial Alzheimer's disease (FAD) cases, in part by increasing the production of Abeta peptides. However, emerging evidence suggests other possible effects of PS1 on synaptic dysfunction where PS1 might contribute to the pathology independent of Abeta. We chose to study the L286V mutation, an aggressive FAD mutation which has never been analyzed at the electrophysiological and morphological levels. In addition, we analyzed for the first time the long term effects of wild-type human PS1 overexpression. We investigated the consequences of the overexpression of either wild-type human PS1 (hPS1) or the L286V mutated PS1 variant (mutPS1) on synaptic functions by analyzing synaptic plasticity and associated spine density changes from 3 to 15 months of age. We found that mutPS1 induces a transient increase observed only in 4- to 5-month-old mutPS1 animals in NMDA receptor (NMDA-R)-mediated responses and LTP compared with hPS1 mice and nontransgenic littermates. The increase in synaptic functions is concomitant with an increase in spine density. With increasing age, however, we found that the overexpression of human wild-type PS1 progressively decreased NMDA-R-mediated synaptic transmission and LTP, without neurodegeneration. These results identify for the first time a transient increase in synaptic function associated with L286V mutated PS1 variant in an age-dependent manner. In addition, they support the view that the PS1 overexpression promotes synaptic dysfunction in an Abeta-independent manner and underline the crucial role of PS1 during both normal and pathological aging.


Assuntos
Envelhecimento , Espinhas Dendríticas/fisiologia , Hipocampo/fisiologia , Plasticidade Neuronal/fisiologia , Neurônios/fisiologia , Presenilina-1/metabolismo , Doença de Alzheimer/genética , Animais , Morte Celular , Espinhas Dendríticas/genética , Modelos Animais de Doenças , Hipocampo/citologia , Humanos , Técnicas In Vitro , Potenciação de Longa Duração/genética , Potenciação de Longa Duração/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto , Plasticidade Neuronal/genética , Neurônios/citologia , Presenilina-1/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Sinapses/genética , Sinapses/fisiologia , Transmissão Sináptica/genética , Transmissão Sináptica/fisiologia
12.
J Clin Med ; 8(9)2019 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-31484320

RESUMO

Mutations in the protein DJ-1 cause autosomal recessive forms of Parkinson's disease (PD) and oxidized DJ-1 is found in the brains of idiopathic PD individuals. While several functions have been ascribed to DJ-1 (most notably protection from oxidative stress), its contribution to PD pathogenesis is not yet clear. Here we provide an overview of the clinical research to date on DJ-1 and the current state of knowledge regarding DJ-1 characterization in the human brain. The relevance of DJ-1 as a PD biomarker is also discussed, as are studies exploring DJ-1 as a possible therapeutic target for PD and neurodegeneration.

13.
Mol Neurobiol ; 56(1): 61-77, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29675578

RESUMO

Mutations in the gene encoding DJ-1 are associated with autosomal recessive forms of Parkinson's disease (PD). DJ-1 plays a role in protection from oxidative stress, but how it functions as an "upstream" oxidative stress sensor and whether this relates to PD is still unclear. Intriguingly, DJ-1 may act as an RNA binding protein associating with specific mRNA transcripts in the human brain. Moreover, we previously reported that the yeast DJ-1 homolog Hsp31 localizes to stress granules (SGs) after glucose starvation, suggesting a role for DJ-1 in RNA dynamics. Here, we report that DJ-1 interacts with several SG components in mammalian cells and localizes to SGs, as well as P-bodies, upon induction of either osmotic or oxidative stress. By purifying the mRNA associated with DJ-1 in mammalian cells, we detected several transcripts and found that subpopulations of these localize to SGs after stress, suggesting that DJ-1 may target specific mRNAs to mRNP granules. Notably, we find that DJ-1 associates with SGs arising from N-methyl-D-aspartate (NMDA) excitotoxicity in primary neurons and parkinsonism-inducing toxins in dopaminergic cell cultures. Thus, our results indicate that DJ-1 is associated with cytoplasmic RNA granules arising during stress and neurodegeneration, providing a possible link between DJ-1 and RNA dynamics which may be relevant for PD pathogenesis.


Assuntos
Grânulos Citoplasmáticos/metabolismo , Degeneração Neural/patologia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Proteína Desglicase DJ-1/metabolismo , Ribonucleoproteínas/metabolismo , Estresse Fisiológico , Animais , Grânulos Citoplasmáticos/efeitos dos fármacos , Células HEK293 , Humanos , Camundongos , N-Metilaspartato/toxicidade , Degeneração Neural/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Pressão Osmótica , Estresse Oxidativo/efeitos dos fármacos , Ligação Proteica , Ratos , Estresse Fisiológico/efeitos dos fármacos
14.
Neurobiol Dis ; 29(3): 465-76, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18201889

RESUMO

TNF-alpha overexpression may contribute to motor neuron death in amyotrophic lateral sclerosis (ALS). We investigated the intracellular pathway associated with TNF-alpha in the wobbler mouse, a murine model of ALS, at the onset of symptoms. TNF-alpha and TNFR1 overexpression and JNK/p38MAPK phosphorylation occurred in neurons and microglia in early symptomatic mice, suggesting that this activation may contribute to motor neuron damage. The involvement of TNF-alpha was further confirmed by the protective effect of treatment with rhTNF-alpha binding protein (rhTBP-1) from 4 to 9 weeks of age. rhTBP-1 reduced the progression of symptoms, motor neuron loss, gliosis and JNK/p38MAPK phosphorylation in wobbler mice, but did not reduce TNF-alpha and TNFR1 levels. rhTBP-1 might possibly bind TNF-alpha and reduce the downstream phosphorylation of two main effectors of the neuroinflammatory response, p38MAPK and JNK.


Assuntos
Esclerose Lateral Amiotrófica/patologia , Esclerose Lateral Amiotrófica/prevenção & controle , Neurônios Motores/patologia , Receptores Tipo I de Fatores de Necrose Tumoral/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Receptores Chamariz do Fator de Necrose Tumoral/uso terapêutico , Fator de Necrose Tumoral alfa/metabolismo , Esclerose Lateral Amiotrófica/genética , Animais , Contagem de Células/métodos , Progressão da Doença , Feminino , Humanos , Masculino , Camundongos , Camundongos Mutantes Neurológicos , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/fisiologia , Receptores Tipo I de Fatores de Necrose Tumoral/administração & dosagem , Proteínas Recombinantes/administração & dosagem , Receptores Chamariz do Fator de Necrose Tumoral/administração & dosagem , Fator de Necrose Tumoral alfa/antagonistas & inibidores
15.
Cerebellum ; 7(4): 534-8, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18949529

RESUMO

In the Lurcher mutant mouse (+/Lc), Purkinje cells (PCs) selectively die due to the mutation that converts alanine to threonine in the glutamate ionotropic receptor GRID 2, thus resulting in a constitutively leaky cation channel. This intrinsic cell death determines a target-dependent cell death of granule cells and olivary neurons and cerebellum cytoarchitecture is severely disrupted in the adult Lurcher mutant. Although the +/Lc mutant has been widely characterized, less is known about the molecules involved in +/Lc PC death. We, here, used organotypic cerebellar slice cultures from P0 mice to investigate the role of c-jun N-terminal kinase (JNK) in +/Lc PC death by using D-JNKI1 as very specific tool to inhibit its action. Our results showed that D-JNKI1 treatment increased the number of +/Lc PC at 14 DIV of 3.6-fold. Conversely, this specific JNK inhibitor cell permeable peptide did not increase PC number in +/+ treated versus untreated cultures. These results clearly indicate that JNK plays an important role in +/Lc PC mechanism of cell death.


Assuntos
Cerebelo/citologia , MAP Quinase Quinase 4/antagonistas & inibidores , Camundongos Mutantes Neurológicos/fisiologia , Peptídeos/fisiologia , Receptores de Glutamato/genética , Alanina/genética , Substituição de Aminoácidos , Animais , Animais Recém-Nascidos , Morte Celular , Permeabilidade da Membrana Celular , Cerebelo/patologia , Cruzamentos Genéticos , Feminino , Genótipo , Masculino , Camundongos , Mutação , Neurônios/enzimologia , Neurônios/fisiologia , Células de Purkinje/citologia , Treonina/genética
16.
Methods Mol Biol ; 399: 1-14, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18309921

RESUMO

To achieve neuroprotection is one of the main interests for neuroscientist: understanding the control mechanisms of neuronal death allows developing new tools for preventing it. Neuronal death plays a critical role in most of the important neural pathologies, including stroke, epilepsy, Parkinson's disease and Alzheimer's disease. This review summarizes the three main different types of neuronal death: apoptosis, necrosis and autophagic cell death, although we are conscious that if cell death falls into several categories, the boundaries are not always distinct. We then introduce the current understanding of the relationship between neuronal death types and neuroprotection.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Encefalopatias/tratamento farmacológico , Encefalopatias/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Animais , Encefalopatias/patologia , Encefalopatias/fisiopatologia , Morte Celular/efeitos dos fármacos , Humanos
17.
Biol Psychiatry ; 82(10): 756-765, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-28187857

RESUMO

BACKGROUND: Kynurenine 3-monooxygenase converts kynurenine to 3-hydroxykynurenine, and its inhibition shunts the kynurenine pathway-which is implicated as dysfunctional in various psychiatric disorders-toward enhanced synthesis of kynurenic acid, an antagonist of both α7 nicotinic acetylcholine and N-methyl-D-aspartate receptors. Possibly as a result of reduced kynurenine 3-monooxygenase activity, elevated central nervous system levels of kynurenic acid have been found in patients with psychotic disorders, including schizophrenia. METHODS: In the present study, we investigated adaptive-and possibly regulatory-changes in mice with a targeted deletion of Kmo (Kmo-/-) and characterized the kynurenine 3-monooxygenase-deficient mice using six behavioral assays relevant for the study of schizophrenia. RESULTS: Genome-wide differential gene expression analyses in the cerebral cortex and cerebellum of these mice identified a network of schizophrenia- and psychosis-related genes, with more pronounced alterations in cerebellar tissue. Kynurenic acid levels were also increased in these brain regions in Kmo-/- mice, with significantly higher levels in the cerebellum than in the cerebrum. Kmo-/- mice exhibited impairments in contextual memory and spent less time than did controls interacting with an unfamiliar mouse in a social interaction paradigm. The mutant animals displayed increased anxiety-like behavior in the elevated plus maze and in a light/dark box. After a D-amphetamine challenge (5 mg/kg, intraperitoneal), Kmo-/- mice showed potentiated horizontal activity in the open field paradigm. CONCLUSIONS: Taken together, these results demonstrate that the elimination of Kmo in mice is associated with multiple gene and functional alterations that appear to duplicate aspects of the psychopathology of several neuropsychiatric disorders.


Assuntos
Quinurenina 3-Mono-Oxigenase/deficiência , Quinurenina 3-Mono-Oxigenase/fisiologia , Transtornos Psicóticos/genética , Transtornos Psicóticos/psicologia , Esquizofrenia/genética , Psicologia do Esquizofrênico , Animais , Cerebelo/metabolismo , Córtex Cerebral/metabolismo , Dextroanfetamina/farmacologia , Ácido Cinurênico/metabolismo , Quinurenina 3-Mono-Oxigenase/genética , Camundongos , Camundongos Knockout , Atividade Motora/efeitos dos fármacos
18.
Adv Exp Med Biol ; 588: 145-55, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17089886

RESUMO

JNKs (c-Jun N- terminal kinases) are important transducing enzymes involved in many faces of cellular regulation such as gene expression, cell proliferation and programmed cell death. The activation of JNK pathway is critical for naturally occurring neuronal death during development as well as for pathological death of adult brain following different insults. In particular, JNKs play an important role in excitotoxicity and all related phenomena. Initial research concentrated on defining the components and organization of JNK signalling cascades, but more recent studies have begun to see JNK as the appropriate target for prevent cell loss. We used a specific JNK inhibitor, the cell permeable peptide D-JNKI1, to block JNK action in neuronal death following excitotoxicity in vitro and cerebral ischemia in vivo. Here we review our recent findings and we discuss the possibility of using D-JNKI1 as a therapeutic agent to prevent cell loss in the central nervous system.


Assuntos
Sistema Nervoso Central/fisiologia , MAP Quinase Quinase 4/fisiologia , Degeneração Neural/patologia , Degeneração Neural/terapia , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/terapia , Animais , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Hipocampo/metabolismo , Hipocampo/patologia , Humanos , Isquemia/patologia , MAP Quinase Quinase 4/metabolismo , Camundongos , Neurônios/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Ratos
19.
J Mol Med (Berl) ; 91(5): 599-611, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23183826

RESUMO

Mutations in the protein DJ-1 cause recessive forms of early onset familial Parkinson's disease (PD). To date, most of the causative mutations studied destabilize formation of DJ-1 homodimers, which appears to be closely linked to its normal function in oxidative stress and other cellular processes. Despite the importance of understanding the dimerization dynamics of this protein, this aspect of DJ-1 biology has not previously been directly studied in living cells. Here, we use bimolecular fluorescence complementation to study DJ-1 dimerization and find not only that DJ-1 forms homodimers in living cells but that most PD causative DJ-1 mutations disrupt this process, including the L166P, M26I, L10P, and P158∆ mutations. Interestingly, the E64D mutant form of DJ-1 retains the ability to form homodimers. However, while wild-type DJ-1 dimers are stabilized under oxidative stress conditions, we find that the E64D mutation blocks this stabilization. Furthermore, our data show that the E64D mutation potentiates the formation of aggresomes containing DJ-1. We also observe that while the widely studied L166P mutation prevents DJ-1 from forming homodimers or heterodimers with wild-type protein, the mutant protein is able to partially disrupt formation of wild-type homodimers. In summary, by investigating DJ-1 dimerization in living cells, we have uncovered several novel properties of PD causative mutations in DJ-1, which may ultimately provide novel insight into PD pathogenesis and possible therapeutic options.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/química , Modelos Moleculares , Mutação , Proteínas Oncogênicas/química , Substituição de Aminoácidos , Expressão Gênica , Vetores Genéticos , Células HEK293 , Humanos , Peróxido de Hidrogênio/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Microscopia Confocal , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Estresse Oxidativo , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Proteína Desglicase DJ-1 , Multimerização Proteica/efeitos dos fármacos , Estabilidade Proteica , Transfecção
20.
Pharmaceuticals (Basel) ; 3(1): 42-58, 2010 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-27713242

RESUMO

The phosphorylation of Amyloid Precursor Protein (APP) at Thr668 plays a key role in APP metabolism that is highly relevant to AD. The c-Jun-N-terminal kinase (JNK), glycogen synthase kinase-3ß (GSK-3ß) and cyclin-dependent kinase 5 (Cdk5) can all be responsible for this phosphorylation. These kinases are activated by excitotoxic stimuli fundamental hallmarks of AD. The exposure of cortical neurons to a high dose of NMDA (100 µM) for 30'-45' led to an increase of P-APP Thr668. During NMDA stimulation APP hyperphosphorylation has to be assigned to GSK-3ß activity, since addition of L803-mts, a substrate competitive inhibitor of GSK-3ß reduced APP phosphorylation induced by NMDA. On the contrary, inhibition of JNK and Cdk5 with D-JNKI1 and Roscovitine respectively did not prevent NMDA-induced P-APP increase. These data show a tight connection, in excitotoxic conditions, between APP metabolism and the GSK-3ß signaling pathway.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA